EP2294183B1 - Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque - Google Patents

Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque Download PDF

Info

Publication number
EP2294183B1
EP2294183B1 EP09767083.0A EP09767083A EP2294183B1 EP 2294183 B1 EP2294183 B1 EP 2294183B1 EP 09767083 A EP09767083 A EP 09767083A EP 2294183 B1 EP2294183 B1 EP 2294183B1
Authority
EP
European Patent Office
Prior art keywords
cells
mscs
tsg
hmscs
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP09767083.0A
Other languages
German (de)
English (en)
Other versions
EP2294183A4 (fr
EP2294183A2 (fr
Inventor
Darwin J. Prockop
Ryang Hwa Lee
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Texas A&M University System
Original Assignee
Texas A&M University System
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Texas A&M University System filed Critical Texas A&M University System
Priority to EP15200119.4A priority Critical patent/EP3029061B1/fr
Publication of EP2294183A2 publication Critical patent/EP2294183A2/fr
Publication of EP2294183A4 publication Critical patent/EP2294183A4/fr
Application granted granted Critical
Publication of EP2294183B1 publication Critical patent/EP2294183B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/25Tumour necrosing factors [TNF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/585Integrins

Definitions

  • the present invention relates to the use of mesenchymal stem cells (also called mesenchymal stromal cells, or MSCs) for repair of cardiac damage and treatment of inflammatory diseases as claimed.
  • mesenchymal stem cells also called mesenchymal stromal cells, or MSCs
  • the compositions used according to the invention may be particularly useful in restoring cardiac function following cardiac damage, including, but not limited to, myocardial infarction, as well as in reducing symptoms of inflammatory disease.
  • the technical problem underlying the present invention was therefore to overcome these prior art difficulties by identifying a suitable cell type for cell-based therapies for heart disease, and by identifying at least one cell-based factor responsible for improved outcomes in heart disease.
  • the solution to this technical problem is provided by the embodiments characterized in the claims.
  • the invention is directed to the use of a population of purified mesenchymal stem cells according to claim 1 or to the use of a population of transgenic mesenchymal stem cells according to claim 3.
  • the inventors have made considerable progress on several of the aforementioned goals as they relate to cell therapy with the stem/progenitors cells from bone marrow, referred to as mesenchymal stem cells or multipotent mesenchymal stromal cells (in either case, "MSCs").
  • the inventors have discovered surface epitopes that define a sub-population of MSCs (defined as RS-MSCs) that demonstrate decreased propensity to generate lethal pulmonary emboli, increased engraftment into infarcted hearts of mice, and more efficient differentiation than preparations of MSCs currently employed by most investigators in the field.
  • RS-MSCs surface epitopes that define a sub-population of MSCs
  • the inventors demonstrate herein that after intravenous (IV) infusion of human MSCs into mice, MSCs entrapped in the lungs are activated to express extremely high levels of a pluripotent anti-inflammatory gene known as "TNF ⁇ stimulated gene 6" (TSG-6).
  • TSG-6 pluripotent anti-inflammatory gene
  • TSG-6 is also known as "tumor necrosis factor-alpha-induced protein 6," or TNFAIP6, and its expression is also induced by interleukin-1 (IL1) and lipopolysaccharide (LPS).
  • IL1 interleukin-1
  • LPS lipopolysaccharide
  • infusions comprising MSCs activated by pre-incubation with TNF ⁇ , IL1, and/or LPS to express elevated levels of TSG-6; 2) infusions comprising MSCs engineered to express elevated levels of TSG-6 (e.g., transfected MSCs over-expressing TSG-6); and 3) infusions comprising recombinant TSG-6 (e.g., recombinant human TSG-6).
  • infusion contemplates both intravenous and intracardiac infusion, unless specifically modified.
  • the invention provides a use of MSCs as claimed in a method of treating cardiac damage comprising administering to a mammal in need thereof a plurality of MSCs, wherein said MSCs are pre-activated MSCs, and further wherein said pre-activated MSCs express elevated levels of TSG-6.
  • the invention also provides a use of MSCs as claimed in a method of treating cardiac damage comprising administering to a mammal in need thereof a plurality of MSCs, wherein said MSCs over-express TSG-6.
  • MSCs as claimed in a method of treating cardiac damage comprising administering to a mammal in need thereof recombinant human TSG-6.
  • the invention additionally provides a use of an MSC as claimed, wherein said MSC has been transfected to over-express TSG-6.
  • the invention further provides the use of MSCs in a pharmaceutically-acceptable preparation of MSCs as claimed, wherein said MSCs have been pre-activated with one or more of the following: TNF ⁇ ; IL1, or LPS.
  • the invention contemplates selecting and isolating TSG-6 positive cells from the population of MSCs pre-activated with one or more cytokines (e.g. so as to isolate an enriched population, i.e. a population with a reduced number of TSG-6 negative cells).
  • positive selection is achieved with an antibody to TSG-6 (e.g. an immobilized antibody for cell capture, a fluorescent antibody for cell sorting, etc.).
  • the invention further provides a use of a purified population of mesenchymal stem cells (MSCs) as claimed that has been contacted with one or more ligands under conditions to express increased levels of TSG-6 protein.
  • the contacting comprises in vitro treatment.
  • the ligand comprises a cytokine, chemokine, and/or LPS.
  • the invention also provides a use of a purified population of mesenchymal stem cells (MSCs) as claimed that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein.
  • the purified population comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the invention also provides the use of a purified population of rapidly self-renewing mesenchymal stem cells (RS-MSCs) that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein.
  • RS-MSCs rapidly self-renewing mesenchymal stem cells
  • the invention further provides the use of a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein as claimed.
  • the population is purified.
  • the population of transgenic mesenchymal stem cells (MSCs) comprises transgenic rapidly self-renewing mesenchymal stem cells (RS-MSCs) that (a) comprise a heterologous nucleotide sequence encoding TSG-6 protein, and (b) express the TSG-6 protein.
  • RS-MSCs transgenic rapidly self-renewing mesenchymal stem cells
  • RS-MSCs transgenic rapidly self-renewing mesenchymal stem cells
  • the invention provides the use in a pharmaceutical composition comprising a population of any of the cells described herein and claimed.
  • the invention further discloses a method for purifying rapidly self-renewing mesenchymal stem cells (RS-MSCs), comprising a) providing a first population of cells comprising RS-MSCs, b) contacting the population of cells with one or both of (i) an antibody that specifically binds to PODXL and (ii) an antibody that specifically binds to CD49f, and c) isolating cells that bind to the one or both of the antibodies, thereby producing a population of purified RS-MSCs.
  • RS-MSCs rapidly self-renewing mesenchymal stem cells
  • the method further comprises d) contacting the population of purified RS-MSCs with one or more of TNF-alpha, IL1, and LPS under conditions to produce a contacted population of cells that expresses increased levels of TSG-6 protein compared to TSG-6 protein levels expressed by the population of purified RS-MSCs.
  • the increased levels of TSG-6 protein are from 10 fold to 500 fold.
  • the method further comprises d) transfecting the population of purified RS-MSCs with a nucleotide sequence that encodes TSG-6 protein.
  • the invention further contemplates a use of a purified population of rapidly self-renewing mesenchymal stem cells (RS-MSCs) produced by any of the methods described in this paragraph.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, and ii) a composition comprising purified tumor necrosis facto-alpha stimulated gene 6 (TSG-6) protein, and b) administering a therapeutically effective amount of the composition to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • administering is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the TSG-6 protein is purified from a transgenic cell that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, and ii) a population of purified mesenchymal stem cells (MSCs) that has been contacted with one or more chemokine, cytokine and LPS under conditions to express increased levels of TSG-6 protein, and b) administering a therapeutically effective amount of the population of purified mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • MSCs mesenchymal stem cells
  • the population of purified mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the step of administering is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the invention provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, ii) a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein, and b)administering a therapeutically effective amount of the population of transgenic mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • MSCs transgenic mesenchymal stem cells
  • the population of transgenic mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the step of administering is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and ii) a population of purified mesenchymal stem cells (MSCs) that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein, and b) administering a therapeutically effective amount of the population of purified mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • MSCs mesenchymal stem cells
  • the population of purified mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the step of administering is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the tissue comprises cardiac muscle tissue, and administering is selected from the group consisting of intramuscular administration into the cardiac muscle tissue and intravenous administration.
  • the subject has, or is at risk of having, a sterile inflammatory disease.
  • a method for reducing one or more symptoms of sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, ii) a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein, and b) administering a therapeutically effective amount of the population of transgenic mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • MSCs transgenic mesenchymal stem cells
  • the population of transgenic mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the tissue comprises cardiac muscle tissue, and administering is selected from the group consisting of intramuscular administration into the cardiac muscle tissue and intravenous administration.
  • the subject has, or is at risk of having, a sterile inflammatory disease.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and ii) a composition comprising purified tumor necrosis facto-alpha stimulated gene 6 (TSG-6) protein, and b) administering a therapeutically effective amount of the composition to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • the tissue comprises cardiac muscle tissue
  • administering is selected from the group consisting of intramuscular administration into the cardiac muscle tissue and intravenous administration.
  • the subject has, or is at risk of having, a sterile inflammatory disease.
  • the TSG-6 protein is purified from a transgenic cell that comprises a heterologous nucleotide sequence encoding TSG-6 protein, and that expresses the TSG-6 protein.
  • the invention additionally provides the use as claimed in a method for detecting sterile inflammation in a tissue in a mammalian subject comprising a) providing a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and b) detecting an increase in serum level of two or more (including three, four, five and six) of plasmin activity, macrophage chemoattractant protein -1 (MCP-1), macrophage inflammatory protein-1 alpha (MIP-1alpha), beta thromboglobulin, soluble ST2 receptor, C-reactive protein (CRP), and natriuretic peptide compared to a control mammalian subject lacking the sterile inflammation in the tissue.
  • the tissue comprises cardiac muscle tissue.
  • the method further comprises c) administering to the subject a therapeutically effective amount of one or more of i) a composition comprising purified tumor necrosis factor-alpha stimulated gene 6 (TSG-6) protein, ii) a population of purified mesenchymal stem cells (MSCs) that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein, and ii) a population of transgenic mesenchymal stem cells (MSCs) that comprises a heterologous nucleotide sequence encoding TSG-6 protein and expresses the TSG-6 protein, wherein the administering produces a treated subject, and d) detecting, in the treated subject, a reduction in serum level of two or more of the plasmin activity, MCP-1, MIP-1alpha, beta thromboglobulin, soluble ST2 receptor, CRP, and natriuretic peptide, compared to the serum level that is detected in step
  • cells and “population of cells” interchangeably refer to a plurality of cells, i.e., more than one cell.
  • the population may be a pure population comprising one cell type. Alternatively, the population may comprise more than one cell type. In the present invention, there is no limit on the number of cell types that a cell population may comprise.
  • MSC Mesenchymal stem cell
  • bone marrow stromal cells bone marrow stromal cells
  • multipotent stromal cell are interchangeably used to refer to a cell derived from bone marrow (reviewed in Prockop, 1997), peripheral blood (Kuznetsov et al., 2001), adipose tissue (Guilak et al., 2004), umbilical cord blood (Rosada et al., 2003), synovial membranes (De Bari et al., 2001), and periodontal ligament (Seo et al., 2005).
  • MSCs are characterized by their ability to adhere to plastic tissue culture surfaces (Friedenstein et al.; reviewed in Owen & Friedenstein, 1988), and by being an effective feeder layers for hematopoietic stem cells (Eaves et al., 2001).
  • MSCs can be differentiated both in culture and in vivo into osteoblasts and chondrocytes, into adipocytes, muscle cells (Wakitani et al., 1995) and cardiomyocytes (Fukuda and Yuasa, 2006), into neural precursors (Woodbury et al., 2000; Deng et al., 2001, Kim et al., 2006; Mareschi et al., 2006; Krampera et al., 2007).
  • MSCs Mesenchymal stem cells
  • MSCs Mesenchymal stem cells
  • Rapidly self-renewing mesenchymal stem cell “RS-MSC” and “type I mesenchymal stem cell” are interchangeably used to refer to an early progenitor cell. They are typically spindle shaped and are present in early-passage MSCs plated at low density. Rapidly self-renewing mesenchymal stem cells (RS-MSCs) may be purified from bone marrow cells and/or from a purified population of mesenchymal stem cells (MSCs) using methods described herein (e.g., binding to one or more of antibody that specifically binds to PODXL, and antibody that specifically binds to CD49f).)
  • SR-MSC slowly replicating mesenchymal stem cell
  • type II mesenchymal stem cell type II mesenchymal stem cell
  • SR-MSC SR-MSC
  • the cells are larger in size than RS-MSC, and are present in early-passage MSCs plated at low density.
  • SR-MSCs arise from RS-MSCs as the cultures expand to confluency.
  • any molecule e.g., amino acid sequence such as PODXL protein, CD49f protein, TSG-6 protein, MCP-1, MIP-1alpha, beta thromboglobulin, soluble ST2 receptor, CRP, natriuretic peptide, antibody that specifically binds to PODXL, antibody that specifically binds to CD49f protein, antibody that specifically binds to TSG-6 protein, etc., and nucleic acid sequence such as those encoding any of the polypeptides described herein), cell (e.g., bone marrow cell, mesenchymal stem cell (MSC), rapidly self-renewing mesenchymal stem cell (RS-MSC), slowly replicating mesenchymal stem cell (SR-MSC), etc.), and/or phenomenon (e.g.,
  • the reduction may be determined subjectively, for example when a patient refers to their subjective perception of disease symptoms, such as pain, fatigue, difficulty in breathing, clarity of vision, nausea, etc.
  • the quantity of molecule, cell, and/or phenomenon in the first sample is lower by any numerical percentage from 5% to 100%, such as, but not limited to, from 10% to 100%, from 20% to 100%, from 30% to 100%, from 40% to 100%, from 50% to 100%, from 60% to 100%, from 70% to 100%, from 80% to 100%, and from 90% to 100% lower than the quantity of the same molecule, cell and/or phenomenon in a second sample.
  • any molecule e.g ., amino acid sequence such as PODXL protein, CD49f protein, TSG-6 protein, MCP-1, MIP-1alpha, beta thromboglobulin, soluble ST2 receptor, CRP, natriuretic peptide, antibody that specifically binds to PODXL, antibody that specifically binds to CD49f protein, antibody that specifically binds to TSG-6 protein, etc., and nucleic acid sequence such as those encoding any of the polypeptides described herein), cell (e.g., bone marrow cell, mesenchymal stem cell (MSC), rapidly self-renewing mesenchymal stem cell (RS-MSC), slowly replicating mesenchymal stem cell (SR-MSC), etc.), and/or phenomenon (e.g., plasmin activity, symptom of a molecule, e. g ., amino acid sequence such as PODXL protein, CD49f protein, TSG-6 protein, MCP-1, MIP-1alpha,
  • the increase may be determined subjectively, for example when a patient refers to their subjective perception of disease symptoms, such as pain, fatigue, difficulty in breathing, clarity of vision, nausea, etc.
  • the quantity of molecule, cell, and/or phenomenon in the first sample is higher by any numerical percentage, such as at least 10% greater than, at least 25% greater than, at least 50% greater than, at least 75% greater than, and/or at least 90% greater than the quantity of the same molecule, cell and/or phenomenon in a second sample.
  • the quantity of molecule, cell, and/or phenomenon in the first sample is higher by any numerical amount from 5 fold to 1000 fold, including from 5 fold to 500 fold, 10 fold to 400 fold, from 20 fold to 300 fold, from 30 fold to 200 fold, from 40 fold to 200 fold, from 50 fold to 200 fold.
  • Cardiac infarction myocardial infarction
  • MI acute myocardial infarction
  • AMI acute myocardial infarction
  • a heart attack which occurs when the blood supply to part of the heart is interrupted causing myocardial muscle cell necrosis. This is most commonly due to occlusion (blockage) of a coronary artery following the rupture of a vulnerable atherosclerotic plaque.
  • ischemia restriction in blood supply
  • oxygen shortage if left untreated for a sufficient period of time, can cause damage and/or death (infarction) of heart muscle tissue (myocardium).
  • treating encompasses delaying and/or reducing the level of one or more objective symptoms and/or one or more subjective symptoms.
  • TSG-6 protein Tumor necrosis factor-alpha stimulated gene 6 protein
  • TNF- ⁇ stimulated gene 6 protein and “TNFAIP6 protein” are used interchangeably to refer to a secretory protein that contains a hyaluronan-binding domain, and thus is a member of the hyaluronan-binding protein family.
  • the hyaluronan-binding domain is known to be involved in extracellular matrix stability and cell migration. This protein has been shown to form a stable complex with inter-alpha-inhibitor (I alpha I), and thus enhance the serine protease inhibitory activity of I alpha I, which is important in the protease network associated with inflammation.
  • I alpha I inter-alpha-inhibitor
  • TSG-6 protein is exemplified by the homo sapiens amino acid sequence of Figure 19A , which is encoded by the nucleotide sequence of Figure 19B (GenBank No. NM_007115). Recombinant purified human TSG-6 protein is commercially available (R&D Systems, Inc., Minneapolis, Catalog # 2104-TS-050).
  • TSG-6 Antibodies that specifically bind to TSG-6 are commercially available (ELISA, monoclonal antibody specific for TSG-6 (clone A38.1.20; Santa Cruz Biotechnology, Inc., Catalog # BAF2104; biotinylated anti-human TSG-6 (TSG-6 Biotinylated PAb Detection Antibody; R&D Systems, Inc., Minneapolis).
  • ELISA monoclonal antibody specific for TSG-6
  • BAF2104 biotinylated anti-human TSG-6
  • TSG-6 Biotinylated PAb Detection Antibody R&D Systems, Inc., Minneapolis
  • PODXL "podocalyxin-like 2,” “endoglycan,” “PODLX2,” “Podocalyxin-like protein 2 precursor,” and “UNQ1861/PRO3742” are interchangeably used, and are exemplified by GenBank Accession no. NM_015720, encoded by mRNA (GenBank) AF219137.
  • Antibodies that specifically bind to PODXL are known in the art, including FITC Labeled anti-Human PCLP1 (Cat.# M084-4, MBL International Corporation, Woburn, Massachusetts).
  • CD49f' "alpha6-integrin” "integrin, alpha 6" and “ITGA6” protein product is the integrin alpha chain alpha 6.
  • Integrins are integral cell-surface proteins composed of an alpha chain and a beta chain. A given chain may combine with multiple partners resulting in different integrins. For example, alpha 6 may combine with beta 4 in the integrin referred to as TSP 180, or with beta 1 in the integrin VLA-6. Integrins are known to participate in cell adhesion as well as cell-surface mediated signaling. Two transcript variants encoding different isoforms have been found for this gene.
  • the amino acid sequence and nucleotide sequence of the exemplary homo sapiens integrin, alpha 6 (ITGA6), transcript variant 2, are described in GenBank Accession No. NM_000210.
  • Homo sapiens chromosome 2, reference assembly, complete sequence is described in GenBank Accession No. NC_000002.11.
  • Antibodies that specifically bind to CD49f are known in the art including PE-Cy5 Rat anti-Human CD49f (Cat.# 551129, BD PharMingen/BD Biosciences.
  • the present invention provides the use of mesenchymal stem cells (MSCs)in the repair of cardiac damage and treatment of inflammatory diseases as claimed.
  • MSCs mesenchymal stem cells
  • the invention also provides the use of TSG-6 protein, that is secreted by MSCs under certain conditions, for repair of cardiac damage and treatment of inflammatory disease.
  • TSG-6 protein that is secreted by MSCs under certain conditions, for repair of cardiac damage and treatment of inflammatory disease.
  • the claimed use of the compositions may be particularly useful in restoring cardiac function following cardiac damage, including, but not limited, to myocardial infarction, as well as in reducing symptoms of inflammatory disease.
  • the inventors disclose herein: (i) the identification of new epitopes that can - for the first time - provide quantitative data on the RS-MSC content of preparations of MSCs; (ii) that RS-MSCs are more clonogenic, have a greater potential to differentiate in culture, and more effective engraft into MI heart and other tissues in mice than the confluent cultures of MSCs (SR-MSCs) employed by most other investigators; (iii) that heart function in MI mice improves after IV infusions of hMSCs because the cells trapped in the lung are activated to secrete large amounts of the multifunctional anti-inflammatory protein TSG-6 that inhibits serine proteinases in the injured heart; and (iv) that some of the protective effects of hMSCs in MI can be reproduced by systemic infusion of rhTSG-6.
  • the invention as claimed is further described under (A) Mesenchymal stromal cells (MSCs), (B) Intravenous hMSCs improve myocardial infarction in mice because cells embolized in the lung are activated to secrete the anti-inflammatory protein TSG-6, (C) Preactivated mesenchymal stem cells (MSCs), and/or preactivated rapidly self-renewing mesenchymal stem cells (RS-MSCs), that overxpress TSG-6 protein, (D) Transgenic mesenchymal stem cells (MSCs), and/or transgenic rapidly self-renewing mesenchymal stem cells (RS-MSCs), that overxpress TSG-6 protein, (E) Pharmaceutical Compositions, (F) Methods for purifying RS-MSCs, (G) Methods for treating cardiac muscle damage by administering preactivated MSCs and/or preactivated RS-MSCs, (H) Methods for treating cardiac muscle damage by administering transgenic MSCs and/or transgenic RS
  • MSCs Mesenchymal stromal cells
  • Bone marrow MSCs have attracted attention in efforts to develop cell therapies (Caplan, 1990;Prockop, 1997; Prockop et al., 2003; Caplan, 2005), because they are readily obtained from patients and expanded in culture.
  • the first clinical trial with MSCs was in patients with severe osteogenesis imperfecta (Horwitz et al., 1999; 2002), a disease of brittle bones caused by mutations in the genes for type I collagen (Prockop, 1985; Prockop & Kivirrikko, 1995).
  • the trial was designed on the basis of data from experiments in a transgenic mouse model developed in our laboratory (Pereira et al., 1998).
  • MSCs originally attracted interest because of their stem-like properties to differentiate into multiple cellular phenotypes, more recent observations presented a paradox: the cells frequently repair injured tissues without much evidence of either engraftment or differentiation.
  • MSCs were used to treat children with severe osteogenesis imperfecta (Horwitz et al., 1999, 2002)
  • children improved in growth rates and other symptoms.
  • assays of tissues from the children revealed that less than 1% of the donor MSCs had engrafted.
  • MSCs provide effective feeder layers for hematopoietic cells (see Eaves et al., 2001), because they secrete a variety of cytokines and chemokines (see Zacharek et al., 2007; Schinkothe et al. 2008; Penolazzi et al., 2007).
  • MSCs respond to cross-talk with injured tissues to enhance repair via a number of different mechanisms, including: 1) enhanced proliferation and differentiation of tissue-endogenous stem/progenitor cells; 2) rescue of ischemic cells by transfer of mitochondria or mitochondrial DNA; 30 suppression of excessive inflammatory responses; and 4) suppression of excessive immune reactions.
  • the inventors also observed that after human MSCs were cocultured with a line of pulmonary epithelial cells with non-functional mitochondria (A549 ⁇ 0 cells), rescued clones of the A549 ⁇ 0 cells with full mitochondrial function were recovered (Spees et al., 2006). Genetic assays indicated that the rescued clones had received mitochondrial DNA from the MSCs without any transfer of genomic DNA or other evidence of cell fusion. Without limiting the invention to a particular mechanism, since loss of functional mitochondria is an early consequence of ischemia, MSCs may in part rescue ischemic injury to myocardium or other tissues since one of the earliest events in ischemic injury is loss of mitochondrial function.
  • Persistent chronic inflammation is now recognized as a contributing factor in a wide variety of diseases ranging from parkinsonism (Tansey et al., 2007; McGeer and McGeer 2007) to diabetes (Theuma and Fonseca 2004; Shoelson et al., 2007).
  • a series of recent reports have emphasized that inflammatory responses to tissue injury in mammals are frequently excessive, and require cell mediators to actively suppress the responses and thereby improve tissue repair (Schwab et al., 2007; Serhan et al., 2008).
  • One class of inflammation suppressors comprises the lipids referred to as lipoxins, resolvins and protectins (Sehran et al., 2008).
  • MSCs provide another mechanism for suppressing inflammation because they can be activated to secrete peptides and proteins that modulate both inflammation and immune responses.
  • Secretion by MSCs of the interleukin 1 (IL1) receptor antagonist apparently explained the improvements observed with administration of MSCs in a lung model of fibrosis induced by bleomycin (Ortiz et al., 2007).
  • Intra-tracheal administration of MSCs suppressed inflammation and prolonged survival of mice by expression of MIP-1 and other cytokines after acute lung inflammation was induced with LPS (Gupta et al., 2007).
  • MSCs suppressed mixed lymphocyte reactions in culture, produced improvements in an animal model for multiple sclerosis (Gerdoni et al., 2007), and improved patients with graft versus host disease (Aggarwal & Pittenger, 2005; Le Blanc & Ringden, 2007).
  • MSCs are activated by IFN ⁇ , together with one of three other pro-inflammatory cytokines to attract T lymphocytes.
  • the MSCs then secrete nitrous oxide to suppress the T lymphocytes.
  • human MSCs injected into the hippocampus of mice after transient global ischemia are activated to reduce neurological deficits and neuron death by suppressing both inflammatory and immune reactions (Ohtaki et al., 2008).
  • MSCs can produce beneficial effects by IV administration both in animal models (Pereira et al., 1998; Akiyama et al., 2002; Chen et al., 2003; Nomura et al., 2005; Wu et al., 2008), and in patients (Horwitz et al., 1999, 2002; Koc et al., 2002; Ringden et al., 2006).
  • the results are surprising, since it has been convincingly demonstrated that most MSCs that are infused IV are rapidly trapped in the lung (Gao et al., 2001; Schrepfer et al., 2007).
  • MSCs in the lung are not in itself unexpected, since it also occurs with polymorphonuclear (PMN)cells (Hogg et al., 1994), metastatic tumors (MacDonald et al., 2002), and probably hematopoietic stem cells (Dooner et al., 2004).
  • PMN polymorphonuclear
  • MacDonald et al., 2002 metastatic tumors
  • Dooner et al., 2004 probably hematopoietic stem cells
  • Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6
  • hMSCs human multipotent stromal cells
  • IV hMSCs but not hMSCs transduced with TSG-6 siRNA decreased inflammatory responses, reduced infarct size, and improved cardiac function.
  • IV administration of recombinant TSG-6 also reduced inflammatory responses and reduced infarct size.
  • the inventors first developed assays to provide quantitative data on the fate of human cells infused into mice. The inventors then demonstrated that IV infused human MSCs (hMSCs) produced functional improvement in mice with myocardial infarction (MI) at least in part because the cells trapped as emboli in lung are activated to express the anti-inflammatory factor TNF- ⁇ induced protein 6 (TNAIP6 or TSG-6).
  • MI myocardial infarction
  • the hMSCs trapped in mouse lung after IV infusion underwent major changes in their patterns of gene expression in response to the injury to the lung produced by micro-embolization of the cells in the pulmonary vasculature (Furlani et al., 2009; Lee et al., 2009).
  • the up-regulation of the human TSG-6 was of special interest because of the anti-inflammatory effects of the protein (Milner et al., 2006; Wisniewski and Vilcek, 2004), and because excessive inflammatory responses contribute to the pathological changes produced by MI (Ovechkin et al., 2005; Paolocci et al., 2006; Carvalho et al., 2006; Fang et al., 2007; Moshal et al., 2008). Therefore the results suggested a possible explanation for the observations that IV infusions of MSCs improved cardiac function in models for MI (Halkos et al., 2008; Iso et al., 2007; Krause et al., 2007; Wolf et al., 2007).
  • TSG-6 The up-regulation of TSG-6 was detected by the cross-species strategy of infusing hMSCs into NOD/scid mice. Similar strategies of using hMSCs in animal models previously proved useful, because the hMSCs provided numerous endogenous markers for the cells and no obvious cross-species artifacts were encountered (Hwang et al., 2008; Lu et al., 2009; Bai et al., 2009; Gonzalez-Rey et al., 2009; Sasportas et al., 2009), apparently because of the immune modulatory effects of the cells (Uccelli et al., 2008).
  • the strategy of using hMSCs avoids the technical difficulties of isolating mouse MSCs (Baddoo et al., 2003; Gnecchi and Melo, 2009; Peister et al., 2004; Sung et al., 2008), and the marked tendency of mouse MSCs to develop genomic instability and become tumorgenic as they are expanded in culture (Sung et al., 2008; Tolar et al., 2007).
  • Permanent LAD ligation in mice does not mimic human MI as closely as ischemia and reperfusion models in larger animals.
  • permanent LAD ligation in NOD/scid mice provided a useful model for testing the effects on hMSCs because the mice retained the excessive inflammatory responses to MI (Iso et al., 2007).
  • TSG-6 is a 30 kDa glycoprotein (Heng et al., 2008; Milner et al., 2006) that was shown to produce three distinct anti-inflammatory effects (Milner et al., 2006; Wisniewski and Vilcek, 2004).
  • TSG-6 may however play a key role in many beneficial effects of MSCs.
  • TSG-6 Inflammatory responses to sterile tissue injury are frequently excessive and require active suppression (Schwab et al., 2007). Also, chronic inflammation plays a key role in diseases such as diabetes, stroke, Alzheimer's disease and parkinsonism (Bergsbaken et al., 2009; McCombe and Read, 2008; Shoelson et al., 2006; Theuma and Fonseca, 2004). Therefore secretion of TSG-6 by MSCs trapped as emboli in lung may in part explain the therapeutic effects observed after IV infusions of MSCs in animal models for these and other diseases (Uccelli et al., 2008; Ezquer et al., 2008; Parr et al., 2007). Secretion of TSG-6 may also play a role in therapies for heart disease with other cells such as skeletal myoblasts, fetal myoblasts and ES cells (Jolicoeur et al., 2007).
  • MSCs Preactivated mesenchymal stem cells
  • RS-MSCs preactivated rapidly self-renewing mesenchymal stem cells
  • the invention provides the use of a purified population of mesenchymal stem cells (MSCs) that has been contacted with one or more ligand selected from the group consisting of pro-inflammatory chemokine, pro-inflammatory cytokine (e.g., TNF-alpha and IL1), and LPS, under conditions to express increased levels of TSG-6 protein.
  • the purified population of mesenchymal stem cells (MSCs) comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • RS-MSCs purified rapidly self-renewing mesenchymal stem cells
  • purified refers to the reduction in the amount of at least one undesirable component (such as cell type, protein, and/or nucleic acid sequence) from a sample, including a reduction by any numerical percentage of from 5% to 100%, such as, but not limited to, from 10% to 100%, from 20% to 100%, from 30% to 100%, from 40% to 100%, from 50% to 100%, from 60% to 100%, from 70% to 100%, from 80% to 100%, and from 90% to 100%.
  • an undesirable component such as cell type, protein, and/or nucleic acid sequence
  • mesenchymal stem cells may be purified from bone marrow cells using methods known in the art (Wakitani et al., 1995; Fukuda and Yuasa, 2006; Woodbury et al., 2000; Deng et al., 2001; Kim et al., 2006; Mareschi et al., 2006; Krampera et al., 2007).
  • rapidly self-renewing mesenchymal stem cells may be purified from bone marrow cells and/or from a purified population of mesenchymal stem cells (MSCs) using methods described herein (e.g., binding to one or more of antibody that specifically binds to PODXL, and/or antibody that specifically binds to CD49f).
  • Cytokine is a category of signaling molecule (protein, peptide, glycoprotein) that is involved in cellular communication.
  • Pro-inflammatory cytokine refers to a cytokine produced predominantly by activated immune cells, such as microglia, and is involved in the amplification of inflammatory reactions.
  • Pro-inflammatory cytokines are exemplified by IL-1alpha, IL-1beta, IL-6, TNF-alpha, and TGF-beta.
  • Other pro-inflammatory mediators include LIF, IFN-gamma, OSM, CNTF, TGF-beta, GM-CSF, IL11, IL12, IL17, IL18, and IL8.
  • Cyclookine and “pro-inflammatory chemokine” interchangeably refer to a molecule that chemoattracts inflammatory cells, and that contains a polypeptide comprising at least two (preferably at least three) cysteine residues that are involved in forming the molecule's 3-dimensional shape.
  • Chemokines include CC chemokines, CXC chemokines, C chemokines, and CX3C chemokines.
  • CC chemokines also referred to as " ⁇ -chemokines” have two adjacent cysteines near their amino terminus, and include those that have four cysteines (C4-CC chemokines), and six cysteines (C6-CC chemokines).
  • CC-chemokine are exemplified by RANTES, AOP-RANTES, CAP-RANTES, HEY-Gly 1 -RANTES, HEA-Gly 1 -RANTES, NNY-RANTES, NNA-RANTES, DDY-RANTES, PSC-RANTES, P1-RANTES, P2-RANTES, C1,C5-RANTES, L-RANTES, Met-RANTES, MIP-1 ⁇ , MIP-1 ⁇ P, AOP-MIP-1 ⁇ , MIP-1ß, vMIP-II.
  • CXC chemokines also referred to as " ⁇ -chemokines”
  • the two N-terminal cysteines are separated by one amino acid (“X").
  • CXC chemokines include chmokines with the motif glutamic acid-Leucine-Arginine (ELR) immediately before the first cysteine of the CXC motif (ELR-positive) (exemplified by CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8), and those without an ELR motif (ELR-negative).
  • ELR glutamic acid-Leucine-Arginine
  • Chemokines with an ELR sequence motif have been found to chemoattract and activate primarily neutrophils.
  • Chemokines without the ELR sequence motif appear to chemoattract and activate monocytes, dendritic cells, T-cells, NK-cells, B-lymphocytes, basophils, and eosinophils.
  • C chemokines also known as “ ⁇ chemokines” have only two cysteines; one N-terminal cysteine and one cysteine downstream.
  • CX 3 C chemokines also known as “ ⁇ -chemokines” have three amino acids between the two cysteines.
  • RS-MSCs isolated preparations of RS-MSCs are far more clonogenic and have greater potential to differentiate in culture than more mature MSCs from more confluent cultures that the inventors have defined as "SM-MSCs" (Smith et al., 2004) and that are currently employed by most investigators.
  • SM-MSCs sub-population of MSCs obtained by serum-starvation of cultures
  • pre-RS-MSCs sub-population of MSCs obtained by serum-starvation of cultures
  • the inventors have defined Dkk-1 derived synthetic peptides that inhibit Wnt signaling and therefore provide a means of recovery and manipulation in culture of early precursor sub-populations of MSCs (Gregory et al., 2005).
  • the inventors have established that the sub-population of MSCs defined as RS-MSCs are engrafted preferentially after IV infusion into immunodeficient mice, and that they migrate more efficiently to cytokines that attract stem cells because of their expression of CXCR4 and CX3R1, the receptors for SDF-1 and fractalkine (Lee et al., 2006).
  • the inventors used ex vivo co-culture experiments to demonstrate that both cell fusion and differentiation without evidence of cell fusion occurred when MSCs were co-cultured with heat-shocked pulmonary epithelial cells or cardiac endothelial cells (Spees et al., 2003).
  • the inventors used ex vivo co-culture experiments to demonstrate the surprising finding that MSCs could rescue cells with non-functional mitochondria by transfer of either intact mitochondria or mitochondrial DNA (Spees et al., 2003).
  • the inventors used experiments with chick embryos to demonstrate that rat MSCs can differentiate into early cardiomyocytes without evidence of cell fusion (Pochampally et al., 2004).
  • the invention provides the use of a purified population of rapidly self-renewing mesenchymal stem cells (RS-MSCs) that has been contacted with one or more of ligands, such as TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein.
  • RS-MSCs rapidly self-renewing mesenchymal stem cells
  • MSCs Transgenic mesenchymal stem cells
  • RS-MSCs transgenic rapidly self-renewing mesenchymal stem cells
  • the invention provides the use of a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein.
  • the population of transgenic mesenchymal stem cells (MSCs) is purified.
  • the population of transgenic mesenchymal stem cells (MSCs) comprises transgenic rapidly self-renewing mesenchymal stem cells (RS-MSCs) that (a) comprise a heterologous nucleotide sequence encoding TSG-6 protein, and (b) express the TSG-6 protein.
  • RS-MSCs transgenic rapidly self-renewing mesenchymal stem cells
  • the invention also provides a population of transgenic rapidly self-renewing mesenchymal stem cells (RS-MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein.
  • RS-MSCs transgenic rapidly self-renewing mesenchymal stem cells
  • transgenic when used in reference to a cell refers to a cell which contains a transgene, or whose genome has been altered by the introduction of a "transgene.”
  • Transgenic cells may be produced by several methods including the introduction of a "transgene” comprising nucleic acid (usually DNA) into a target cell or integration of the transgene into a chromosome of a target cell by way of human intervention, using methods known in the art such as vectors (e.g., plasmids, linear DNA, encapsidated virus, etc.)
  • transgene refers to any nucleic acid sequence that is introduced into the cell by experimental manipulations.
  • a transgene may be an "endogenous DNA sequence” or a “heterologous DNA sequence.”
  • endogenous DNA sequence refers to a nucleotide sequence that is naturally found in the cell into which it is introduced so long as it does not contain some modification (e.g. , a point mutation, the presence of a selectable marker gene, etc.) relative to the naturally occurring sequence.
  • heterologous DNA sequence and “foreign DNA sequence" interchangeably refer to a nucleotide sequence that is ligated to, or is manipulated to become ligated to, a nucleic acid sequence to which it is not ligated in nature, or to which it is ligated at a different location in nature.
  • Heterologous DNA is not endogenous to the cell into which it is introduced, but has been obtained from another cell.
  • Heterologous DNA also includes an endogenous DNA sequence that contains some modification.
  • heterologous DNA encodes RNA and proteins that are not normally produced by the cell into which it is expressed. Examples of heterologous DNA include reporter genes, transcriptional and translational regulatory sequences, selectable marker proteins ( e.g. , proteins which confer drug resistance), etc.
  • Vectors i.e., plasmids, linear DNA, encapsidated virus, etc. may be introduced into cells using techniques well known in the art.
  • the term "introducing" a nucleic acid sequence into a cell refers to the introduction of the nucleic acid sequence into a target cell to produce a "transformed” or “transgenic” cell. Methods of introducing nucleic acid sequences into cells are well known in the art.
  • the sequence may be "transfected" into the cell using, for example, calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, and biolistics.
  • the sequence may be introduced into a cell by "infecting" the cell with the virus.
  • Transformation of a cell may be stable or transient.
  • transient transformation and “transiently transformed” refer to the introduction of one or more nucleotide sequences of interest into a cell in the absence of integration of the nucleotide sequence of interest into the host cell's genome.
  • Transient transformation may be detected by, for example, enzyme-linked immunosorbent assay (ELISA) that detects the presence of a polypeptide encoded by one or more of the nucleotide sequences of interest.
  • ELISA enzyme-linked immunosorbent assay
  • transient transformation may be detected by detecting the activity of the protein encoded by the nucleotide sequence of interest.
  • transient transformant refer to a cell that has transiently incorporated one or more nucleotide sequences of interest.
  • stable transformation and “stably transformed” refer to the introduction and integration of one or more nucleotide sequence of interest into the genome of a cell.
  • a “stable transformant” is distinguished from a transient transformant in that, whereas genomic DNA from the stable transformant contains one or more heterologous nucleotide sequences of interest, genomic DNA from the transient transformant does not contain the heterologous nucleotide sequence of interest.
  • Stable transformation of a cell may be detected by Southern blot hybridization of genomic DNA of the cell with nucleic acid sequences that are capable of binding to one or more of the nucleotide sequences of interest.
  • stable transformation of a cell may also be detected by the polymerase chain reaction of genomic DNA of the cell to amplify the nucleotide sequence of interest.
  • Gene expression refers to the process of converting genetic information encoded in a gene into RNA (e.g., mRNA, rRNA, tRNA, or snRNA) through “transcription” of the gene (i.e., via the enzymatic action of an RNA polymerase), and for protein encoding genes, into protein through “translation” of mRNA.
  • Gene expression can be regulated at many stages in the process.
  • Up-regulation” or “activation” refers to regulation that increases the production of gene expression products (i.e., RNA or protein), while “down-regulation” or “repression” refers to regulation that decrease production.
  • Molecules e.g., transcription factors
  • activators e.g., transcription factors
  • the invention additionally provides the use of a pharmaceutical composition
  • a pharmaceutical composition comprising the purified MSCs, and/or purified RS-MSCs, and/or transgenic MSCs that express TSG-6, and/or transgenic RS-MSCs that express TSG-6, and/or purified TSG-6.
  • pharmaceutically acceptable molecules i.e., molecules that are capable of administration to or upon a subject and that do not substantially produce an undesirable effect such as, for example, adverse or allergic reactions, dizziness, gastric upset, toxicity and the like, when administered to a subject.
  • the pharmaceutically acceptable molecule does not substantially reduce the activity of the invention's compositions.
  • Pharmaceutical molecules include, but are not limited to, excipients and diluents.
  • Excipient is an inactive substance used as a carrier for the invention's compositions that may be useful for delivery, absorption, bulking up to allow for convenient and accurate dosage of the invention's compositions.
  • Excipients include, without limitation, antiadherents, binders (e.g., starches, sugars, cellulose, modified cellulose such as hydroxyethyl cellulose, hydroxypropyl cellulose and methyl cellulose, lactose, sugar alcohols such as xylitol, sorbital and maltitol, gelatin, polyvinyl pyrrolidone, polyethylene glycol), coatings (e.g., shellac, corn protein zein, polysaccharides), disintegrants (e.g., starch, cellulose, crosslinked polyvinyl pyrrolidone, sodium starch glycolate, sodium carboxymethyl cellulosemethycellulose), fillers (e.g., cellulose, gelatin, calcium phosphate, vegetable fats and oils, and sugars
  • the excipient comprises HEC (hydroxyethylcellulose), which is a nonionic, water-soluble polymer that can thicken, suspend, bind, emulsify, form films, stabilize, disperse, retain water, and provide protective colloid action.
  • HEC hydroxyethylcellulose
  • Exemplary "diluents” include water, saline solution, human serum albumin, oils, polyethylene glycols, aqueous dextrose, glycerin, propylene glycol or other synthetic solvents.
  • the invention discloses a method for purifying rapidly self-renewing mesenchymal stem cells (RS-MSCs), comprising a) providing a first population of cells comprising RS-MSCs, b) contacting the population of cells with one or both of (i) an antibody that specifically binds to PODXL and (ii) an antibody that specifically binds to CD49f, and c) isolating cells that bind to the one or both of the antibodies, thereby producing a population of purified RS-MSCs.
  • RS-MSCs rapidly self-renewing mesenchymal stem cells
  • Fig 3 shows the changes in the epitopes in MSCs with expansion in culture, and that cell populations enriched for PODXL hi /CD49F hi cells demonstrate increased clonogenicity and differentiation potential compared to the MSC cells population from which they are isolated.
  • antibody encompasses any immunoglobulin (e.g. , IgG, IgM, IgA, IgE, IgD, etc. ) obtained from any source (e.g., humans, rodents, non-human primates, caprines, bovines, equines, ovines, etc.). Included within this definition are polyclonal antibody, monoclonal antibody, and chimeric antibody. Methods for making monoclonal antibodies are known (See e.g.
  • binding specificity refers to the preferential interaction between the first molecule with the second molecule as compared to the interaction between the second molecule with a third molecule.
  • Specific binding is a relative term that does not require absolute specificity of binding; in other words, the term “specific binding” does not require that the second molecule interact with the first molecule in the absence of an interaction between the second molecule and the third molecule.
  • the level of interaction between the first molecule and the second molecule is higher than the level of interaction between the second molecule with the third molecule.
  • "Specific binding" of a first molecule with a second molecule also means that the interaction between the first molecule and the second molecule is dependent upon the presence of a particular structure on or within the first molecule. For example, if a second molecule is specific for structure "A" that is on or within a first molecule, the presence of a third nucleic acid sequence containing structure A will reduce the amount of the second molecule which is bound to the first molecule.
  • Antibodies that specifically bind to PODXL are known in the art, including FITC Labeled anti-Human PCLP1 (Cat.# M084-4, MBL International Corporation, Woburn, Massachusetts).
  • Antibodies that specifically bind to CD49f are known in the art including PE-Cy5 Rat anti-Human CD49f (Cat.# 551129, BD PharMingen/BD Biosciences.
  • the methods further include step d) contacting the population of purified RS-MSCs with one or more of TNF-alpha, IL1, and LPS under conditions to produce a contacted population of cells that expresses increased levels of TSG-6 protein compared to TSG-6 protein levels expressed by the population of purified RS-MSCs.
  • the increased levels of TSG-6 protein are from 10 fold to 500 fold.
  • the increased levels of TSG-6 protein is higher by any numerical amount from 5 fold to 1000 fold, including from 5 fold to 500 fold, 10 fold to 400 fold, from 20 fold to 300 fold, from 30 fold to 200 fold, from 40 fold to 200 fold, from 50 fold to 200 fold.
  • the methods comprise step d) transfecting the population of purified RS-MSCs with a nucleotide sequence that encodes TSG-6 protein (e.g., sequences of Figure 19 ).
  • the invention further contemplates a purified population of rapidly self-renewing mesenchymal stem cells (RS-MSCs) produced by the methods described herein.
  • the invention provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, and ii) a population of purified mesenchymal stem cells (MSCs) that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein, and b) administering a therapeutically effective amount of the population of purified mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • the population of purified mesenchymal stem cells (MSCs) comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • Subject and “animal” interchangeably refer to any multicellular animal, preferably a mammal, e.g. , humans, non-human primates, murines, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, canines, felines, aves, etc. ).
  • mammalian subjects include mouse, rat, guinea pig, hamster, ferret and chinchilla.
  • Subject in need of reducing one or more symptoms of includes a subject that exhibits and/or is at risk of exhibiting one or more symptoms of the disease.
  • subjects may be at risk based on family history, genetic factors, environmental factors, etc. This term includes animal models of the disease, such as the mouse models described herein.
  • the terms “therapeutically effective amount” and “protective amount” of a composition with respect to cardiac muscle cell necrosis and/or myocardial infarction, interchangeably refer to, in one embodiment, an amount of the composition that delays, reduces, palliates, ameliorates, stabilizes, prevents and/or reverses one or more symptoms of the disease compared to in the absence of the composition of interest. It is not necessary that all symptoms be completely eliminated.
  • the term “delaying” symptoms refers to increasing the time period during which symptoms are detectable.
  • the term “eliminating” symptoms refers to 100% reduction of one or more symptoms.
  • a pharmaceutically effective amount may be determined using in vitro and in vivo assays known in the art and disclosed herein, as well as clinical trials. The amount depends, for example, on the route of administration, patient weight (e.g. milligrams of drug per kg body weight). These factors and their relationship to determining this amount are well known to skilled practitioners in the medical, veterinary, and other related arts. This amount and the method of administration can be tailored to achieve optimal efficacy but will depend on such factors as weight, diet, concurrent medication and other factors that those skilled in the art will recognize. The dosage amount and frequency are selected to create an effective level of the composition without substantially harmful effects.
  • the dosage of the polypeptides will generally range from 0.001 to 1000 mg/Kg/day, more preferably from 0.01 to 100 mg/Kg/day, and most preferably from 0.1 to 10 mg/Kg/day.
  • Symptoms of myocardial muscle cell necrosis and “symptoms of cardiac infarction” are used to refer to objective and/or subjective symptoms.
  • Objective symptoms are exemplified by increased plasmin activity in serum ( Fig. 9A ), increased cardiac tissue levels of one or more of plasminogen activator (tPA), urokinase (uPA), pro-matrix metalloproteinase 9 (pro-MMP9), active MMP9 ( Figs. 9B and 9C ), and cardiac muscle cell death.
  • Symptoms may also include subjective symptoms such as chest pain, shortness of breath, nausea, vomiting, palpitations, sweating, anxiety, feeling of indigestion, fatigue, etc.
  • Necrosis refers to the premature death of cells and living tissue. Necrosis is typically caused by external factors, such as infection, toxins, trauma, reduced blood supply, etc. This is in contrast to apoptosis, which is a naturally occurring cause of cellular death.
  • administering refers to introducing a polypeptide, introducing a nucleic acid sequence encoding a polypeptide, and/or introducing a host cell that expresses a polypeptide.
  • Polypeptides may be administered to a subject using methods known in the art (e.g., Erickson et al., U.S. Patent 6,632,979 ; Furuta et al., U.S. Patent 6,905,839 ; Jackobsen et al., U.S. Patent 6,238,878 ; Simon et al., U.S. Patent 5,851,789 ), including those for administering bacteria ( Bellinger et al., U.S.
  • Patent 6,964,856 and for administering antisense ( de la Monte et al., U.S. Patent7,291,454 ; Smith et al., WO 90/09180 ; Squinto et al., WO 93/00909 ), and oligonucleotides ( Inoyue et al., U.S. Patent 5,272,065 ).
  • the polypeptides, nucleic acid sequences and/or cells may be administered prophylactically (i.e., before the observation of disease symptoms) and/or therapeutically (i.e., after the observation of disease symptoms). Administration also may be concomitant with (i.e., at the same time as, or during) manifestation of one or more disease symptoms.
  • the invention's compositions may be administered before, concomitantly with, and/or after administration of another type of drug or therapeutic procedure (e.g. , surgery).
  • Methods of administering the invention's compositions include, without limitation, administration in parenteral, oral, intraperitoneal, intranasal, topical ( e.g. , rectal, and vaginal), and sublingual forms.
  • Parenteral routes of administration include, for example, subcutaneous, intravenous, intramuscular, intrastemal injection, and infusion routes.
  • the route of administration is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the invention provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, ii) a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein, and b) administering a therapeutically effective amount of the population of transgenic mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • the population of transgenic mesenchymal stem cells (MSCs) comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the route of administration is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of cardiac muscle cell necrosis in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of cardiac muscle cell necrosis, and ii) a composition comprising purified tumor necrosis factor-alpha stimulated gene 6 (TSG-6) protein, and b) administering a therapeutically effective amount of the composition to the mammalian subject, thereby reducing one or more symptoms of the cardiac muscle cell necrosis.
  • TSG-6 tumor necrosis factor-alpha stimulated gene 6
  • TSG-6 protein Purified recombinant TSG-6 protein, is commercially available (R&D Systems, Inc., Minneapolis, Catalog # 2104-TS-050).
  • TSG-6 protein may be purified from a transgenic cell that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein.
  • any cell that may be transformed to express a heterologous nucleotide sequence may be used to express TSG-6 protein.
  • Such cells include human and non-human eukaryotic animal cells.
  • the cell is a human eukaryotic animal cell as exemplified by U937 cells (macrophage), ATCC# crl 1593.2; A-375 cells (melanoma/melanocyte), ATCC# crl-1619; KLE cells (uterine endometrium), ATCC# crl-1622; T98G cells (glioblastoma), ATCC# crl-1690; CCF-STTG1 cells (astrocytoma), ATCC# crl-1718; HUV-EC-C cells (vascular endothelium), ATCC# CRL-1730; UM-UC-3 cells (bladder), ATCC# crl-1749; CCD841-CoN cells (colon, ATCC# crl-1790; SNU-423 cells (hepatocellular carcinoma), ATCC# crl-2238;
  • the cell is a non-human eukaryotic animal cell exemplified by, but not limited to, yeast cells (AH109), LM cells (mouse fibroblast), ATCC# ccl-1.2; NCTC 3526 cells (rhesus monkey kidney), ATCC# ccl-7.2; BHK-21 cells (golden hamster kidney), ATCC# ccl-10 ; MDBK cells (bovine kidney), ATCC# ccl-22; PK 15 cells (pig kidney), ATCC# ccl-33; MDCK cells (dog kidney), ATCC# ccl-34; PtK1 cells (kangaroo rat kidney), ATCC# ccl-35; Rk 13 cells (rabbit kidney), ATCC# ccl-37; Dede cells (Chinese hamster lung fibroblast), ATCC# ccl-39; Bu (IMR31) cells (bison lung fibroblast), ATCC# ccl-40; FHM cells (minnow
  • NBL-6 cells horse fibroblast
  • ATCC# ccl-57 MvLn cells
  • ATCC# ccl-64 Ch1 Es cells (goat fibroblast), ATCC# ccl-73; P1 I Nt cells (raccoon fibroblast), ATCC# ccl-74; Sp I k cells (dolphin epithelial), ATCC# ccl-78; CRFK cells (cat epithelial), ATCC# ccl-94; Gekko Lung 1 cells (lizard-gekko epithelial), ATCC# ccl-111; Aedes Aegypti cells (mosquito epithelial), ATCC# ccl-125; ICR 134 cells (frog epithelial), ATCC# ccl-128; Duck embryo cells (duck fibroblast), ATCC# ccl-141; DBS Fcl-1 cells (monkey lung fibro
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of inflammation, including, but not limited to, sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and ii) a population of purified mesenchymal stem cells (MSCs) that has been contacted with one or more of TNF-alpha, IL1, and LPS under conditions to express increased levels of TSG-6 protein, and b) administering a therapeutically effective amount of the population of purified mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • MSCs mesenchymal stem cells
  • the population of purified mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the subject has, or is at risk of having, a sterile inflammatory disease. Inflammation can also be due to an autoimmune response.
  • Inflammation "inflammatory" and grammatical equivalents when in reference to a disease refer to the complex biological response of vascular tissues to harmful stimuli, such as pathogens, damaged cells, or irritants. It is a protective attempt by the organism to remove the injurious stimuli as well as initiate the healing process for the tissue.
  • Inflammation can be acute or chronic.
  • Acute inflammation is the initial response of the body to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood into the injured tissues.
  • a cascade of biochemical events propagates and matures the inflammatory response, involving the local vascular system, the immune system, and various cells within the injured tissue.
  • Prolonged inflammation known as chronic inflammation, leads to a progressive shift in the type of cells which are present at the site of inflammation and is characterized by simultaneous destruction and healing of the tissue from the inflammatory process.
  • Inflammation is not a synonym for infection by a pathogen.
  • sterile inflammation refers to inflammation that is not caused by a pathogen (e.g., bacteria, virus, etc.), but which is caused in response to an injury or abnormal stimulation caused by a physical, chemical, or biologic molecule (protein, DNA, etc.); these reactions include the local reactions and resulting morphologic changes, destruction or removal of the injurious material, and responses that lead to repair and healing.
  • pathogen e.g., bacteria, virus, etc.
  • proteins protein, DNA, etc.
  • TCR T-cell receptor
  • Sterile inflammatory disease and conditions may be systemic (e.g., lupus) or localized to particular tissues or organs.
  • sterile inflammatory diseases include, without limitation, myocardial infarction (MI), diabetes, stroke, Alzheimer's disease, multiple sclerosis, parkinsonism, nephritis, cancer, inflammatory diseases involving acute or chronic inflammation of bone and/or cartilage in a joint, anaphylactic reaction, asthma, conjunctivitis, systemic lupus erythematosus, pulmonary sarcoidosis, ocular inflammation, allergy, emphysema, ischemia-reperfusion injury, fibromyalagia, and inflammatory cutaneous disease selected from psoriasis and dermatitis, or an arthritis selected from rheumatoid arthritis, gouty arthritis, juvenile rheumatoid arthritis, and osteoarthritis.
  • MI myocardial infarction
  • diabetes diabetes
  • stroke Alzheimer's disease
  • multiple sclerosis multiple sclerosis
  • parkinsonism nephritis
  • cancer inflammatory diseases involving acute or
  • Symptoms of inflammation and “symptoms of sterile inflammation” are interchangeably used to refers to objective and/or subjective symptoms.
  • Objective symptoms are exemplified by increased serum levels of one or more (including two, three, four, five, six, and seven) of plasmin activity (Example 11, Figs. 9 and 16 ), macrophage chemoattractant protein -1 (MCP-1), macrophage inflammatory protein-1 alpha (MIP-1alpha), beta thromboglobulin, soluble ST2 receptor, C-reactive protein (CRP), and natriuretic peptide compared to a control mammalian subject lacking inflammation in the tissue.
  • Subjective symptoms include pain.
  • the route of administration is selected from the group consisting of intramuscular administration into cardiac muscle and intravenous administration.
  • the invention also provides the use as claimed in a method for reducing one or more symptoms of sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, ii) a population of transgenic mesenchymal stem cells (MSCs) that (a) comprises a heterologous nucleotide sequence encoding TSG-6 protein, and (b) expresses the TSG-6 protein, and b)administering a therapeutically effective amount of the population of transgenic mesenchymal stem cells (MSCs) to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • MSCs transgenic mesenchymal stem cells
  • the population of transgenic mesenchymal stem cells comprises purified rapidly self-renewing mesenchymal stem cells (RS-MSCs).
  • the tissue comprises cardiac muscle tissue and the route of administration is selected from the group consisting of intramuscular administration into the cardiac muscle tissue and intravenous administration.
  • the subject has, or is at risk of having, a sterile inflammatory disease.
  • the invention further provides the use as claimed in a method for reducing one or more symptoms of sterile inflammation in a mammalian subject comprising a) providing i) a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and ii) a composition comprising purified tumor necrosis factor-alpha stimulated gene 6 (TSG-6) protein, and b) administering a therapeutically effective amount of the composition to the mammalian subject, thereby reducing one or more symptoms of the sterile inflammation in the tissue.
  • the tissue comprises cardiac muscle tissue and the route of administration is selected from the group consisting of intramuscular administration into the cardiac muscle tissue and intravenous administration.
  • the subject has, or is at risk of having, a sterile inflammatory disease.
  • the TSG-6 protein is purified from a transgenic cell that comprises a heterologous nucleotide sequence encoding TSG-6 protein, and that expresses the TSG-6 protein.
  • the invention additionally provides the use as claimed in methods for detecting sterile inflammation in a tissue in a mammalian subject comprising a) providing a mammalian subject in need of reducing one or more symptoms of sterile inflammation in a tissue, and b) detecting an increase in serum level of one or more (including two, three, four, five, six and seven) of plasmin activity, macrophage chemoattractant protein -1 (MCP-1), macrophage inflammatory protein- 1 alpha (MIP-I alpha), beta thromboglobulin, soluble ST2 receptor, C-reactive protein (CRP), , and natriuretic peptide compared to a control mammalian subject lacking the sterile inflammation in the tissue.
  • the tissue comprises cardiac muscle tissue.
  • the levels of the biomarkers used in the invention's methods may be determined using standard techniques. For example, methods for detection of plasmin activity are described herein (Example 11, Figs. 9 and 16 ); MCP-I and MIP-I alpha may be assayed using a commercial ELISA kit from Leinco Technologies, Inc., St. Louis, Missouri; Beta thromboglobulin may be assayed using the commercial ELISA kit (ASSERACHROM B-TG) from DIAGNOSTICA STAGO, Inc., Parsippany, NJ; ST2 may be assayed using the commercial ELISA kit from MBL, Woburn, MA, and CRP may be assayed by Quest Diagnostics.
  • MCP-I Macrophage chemoattractant protein -1
  • MIP-1alpha macrophage inflammatory protein-1 alpha
  • beta thromboglobulin ( Riza et al. (2004) Coron Artery Dis.; 15:265-8 ); soluble ST2 receptor ( Weinberg et al. (2002) Circulation 106:2961-2966 ), C-reactive protein (CRP) ( Pye et al. (1990) Br Heart J 63:228-230 ); and natriuretic peptide ((Weinberg et al. (2002).
  • the method further comprises c) administering to the subject a therapeutically effective amount of one or more of i)a composition comprising purified tumor necrosis factor-alpha stimulated gene 6 (TSG-6) protein, ii) a population of purified mesenchymal stem cells (MSCs) of Claim 1, and ii) a population of transgenic mesenchymal stem cells (MSCs) of Claim 4, wherein the administering produces a treated subject, and d)detecting, in the treated subject, a reduction in serum level of two or more of the plasmin activity, MCP-1, MIP-1alpha, beta thromboglobulin, soluble ST2 receptor, CRP, and natriuretic peptide compared to the serum level that is detected in step b).
  • TSG-6 tumor necrosis factor-alpha stimulated gene 6
  • MSCs mesenchymal stem cells
  • MSCs transgenic mesenchymal stem cells
  • FIGS. 1A and 1B the inventors searched for antibodies to surface proteins that identify early progenitors in cultures of MSCs.
  • FIGS. 1A and 1B the inventors queried microarray data for changes in transcripts for surface proteins as hMSCs plated at low density and the cultures expanded.
  • MSCs were plated at 100 cells/cm 2 .
  • the cultures were expanded for 5 days to obtain MSCs that were PODXL hi /CD49f hi and for 9 days to obtain MSCs that were PODXL lo /CD49f lo .
  • Two sub-populations were then re-plated at 1 cell/cm 2 to assay colony forming units (CFU-Fs).
  • cultures of MSCs from some donors remained PODXL hi /CD49f hi when plated at 100 cells/cm 2 and expanded for up to 7 days.
  • 7 day cultures enriched for RS-MSCs from such selected donors were used for subsequent experiments. Unless otherwise noted, they are referred to simply as MSCs.
  • Alu sequences were assayed in blood, lung and seven tissues of mice for up to 4 days (100 hr) after IV infusions ( FIG. 6 , Top). There was no increase in human cells circulating in blood in the first 60 min, suggesting that few of the trapped cells were leaving the lung. Also, the recovery of the infused human DNA in the seven other tissues was less than a total of 0.2%, or fewer than 2,000 cells.
  • the assays for Alu sequences in lung reflected live MSCs, since essentially same values were obtained by assays for human GAPDH mRNA ( FIG. 6 , Top Left). The number of MSCs trapped was decreased by immediate pretreatment of the mice with a vasodilator (sodium nitroprusside) ( FIG.
  • transcriptomes of both mouse lung cells and hMSCs change after hMSCs are trapped in the lung
  • TNF- ⁇ stimulated gene 6 TNF- ⁇ stimulated gene 6
  • TSG-6 TNF- ⁇ stimulated gene 6
  • the 30 kDa protein was demonstrated to reduce inflammation by several actions: (i) it binds to and blocks the pro-inflammatory effects of fragments of hyaluronan; (ii) it forms a stable complex with inter- ⁇ -inhibitor and thus produces a 100-fold increase in the inhibition of serine proteases that are essential components of most inflammatory responses; (iii) it inhibits neutrophil chemotaxis; and (iv) in animal models of arthritis, it protects joint cartilage from degradation.
  • hMSCs can be activated to express high levels of TSG-6
  • hMSCs were incubated with the pro-inflammatory cytokine TNF- ⁇ .
  • the hMSCs were activated to express 60- to 120-fold levels of the TSG-6 transcript ( FIG. 8 , Top Left).
  • Unstimulated hMSCs did not secrete measurable amounts of the protein but hMSCs activated by TNF- ⁇ secreted large amounts ( FIG. 8 , Middle Panels).
  • Knock down of the gene with siRNAs decreased both the level of the transcript and the secretion of the protein ( FIG. 8 , Bottom Panels).
  • GPRIN3 GPRIN family member 3 4.783493 235410_at NPHP3: nephronophthisis 3 (adolescent) 4.783217 211170_s_at PDE10A: phosphodiesterase 10A 4.778784 233770_at CDNA FLJ12077 fis, clone HEMBB1002453 4.754414 224862_at GNAQ: Guanine nucleotide binding protein (G protein), q polypeptide 4.628492 202738_s_at PHKB: phosphorylase kinase, beta 4.616261 218130_at C17orf62: chromosome 17 open reading frame 62 4.565367 213664_at SLC1A1: solute carrier family 1 (neuronal/epithelial high affinity glutamate transporter, system Xag), member 1 4.554664 1556768 at CDNA FLJ35829 fis,
  • tissue-specific standard curves minimized variations introduced by differences in yields of extracted DNA, cell numbers of the organs, or efficiencies of the PCR reactions.
  • the sensitivity of the assay was about 100 human cells per mouse organ assayed.
  • a quantitative colorimetric assay for DNA in extracts (BURTON, 1956) was used instead of UV absorbance to select appropriate aliquots for the PCR reactions.
  • BURTON a quantitative colorimetric assay for DNA in extracts
  • the fraction of hMSCs trapped in the lung was not significantly reduced by decreasing the number of infused MSCs to a little as 10,000 in the same volume of vehicle (150 ⁇ l), pre-treating the cells with antibodies to integrin- ⁇ 4 or integrin- ⁇ 6 (Qian et al., 2006), or pre-incubating the cells with rat white blood cells (Chute, 2006).
  • 2 X 10 6 hMSCs were infused into the left ventricle of the heart.
  • hMSCs were infused into the tail veins for NOD/scid mice 1 day after MIs were produced by permanent ligation of the anterior descending coronary artery (LAD).
  • LAD anterior descending coronary artery
  • Assays for Alu sequences indicated that 0.04 % +/- 0.03 SD of the infused cells (400 cells +/- 300 SD; n 5) were recovered in the infracted hearts 15 min after the infusion ( Figure 13F ).
  • the up-regulated 451 human transcripts were subjectively examined for candidate genes of interest and human-specific real-time RT-PCR assays were used to confirm the microarray data ( Figure 15A ).
  • the results confirmed 2-fold or greater increases in the transcripts for SMAD6, CSF1, VCAM-1 and TNFAIP6 (TSG-6).
  • the increases in TSG-6 were 28-fold and 47-fold or considerably larger than the 7.5-fold increase detected by the microarrays (Supplemental Table 1).
  • real-time RT-PCR assays frequently demonstrated larger changes in transcripts than microarray assays with the system employed here.
  • TSG-6 The increase in TSG-6 was of particular interest because the protein was previously shown to be a powerful anti-inflammatory factor (Forteza et al., 2007; Getting et al., 2002; Wisniewski and Vilcek, 2004; Milner et al., 2006).
  • Real-time RT-PCR assays demonstrated that human TSG-6 mRNA in lung was increased at 10 hr and further increased at 24 hr after IV infusions of hMSCs ( Figure 15B ). There was no difference in expression of TSG-6 in lungs from naive mice and mice with MIs ( Figure 15B ).
  • TSG-6 was discovered by analysis of cDNA clones from skin fibroblasts that were incubated with TNF- ⁇ (Lee et al., 1992). Therefore, hMSCs and fibroblasts from same donor were incubated with TNF- ⁇ and the mRNAs were assayed by real-time RT-PCR.
  • the transcript for TSG-6 in hMSCs was increased about 120-fold after incubation with 10 ng/ml TNF- ⁇ for 48 hr and increased about 80-fold with a further passage of the hMSCs ( Figure 15C ).
  • TSG-6 siRNA Transient transduction of hMSCs with TSG-6 siRNA abrogated the effects of TNF- ⁇ on TSG-6 transcription ( Figure 15 E) and secretion ( Figure 3 F). Expression of TSG-6 was partially reduced by a mock transduction or transduction with a scrambled siRNA.
  • Acute MI produces an acute inflammatory response in which infiltrating neutrophils generate MMPs that degrade the myocardium (Fang et al., 2007; Lindsey et al., 2001).
  • the permanent LAD increased serum levels of both cardiac troponin I ( Figure 16A ), a biomarker for myocardial injury (Chapelle, 1998; Pervaiz et al., 1997), and plasmin activity ( Figure 16B ), a marker for inflammatory responses (Heymans et al., 1999; Griffin et al., 2005).
  • the plasmin activity was decreased by two infusions of rhTSG-6, an observation consistent with its known inhibitory effects (Bardos et al., 2001; Milner et al., 2006).
  • the plasmin activity was also decreased by IV infusion of hMSCs and hMSCs with a scrambled siRNA but not hMSCs transduced with siRNA for TSG-6.
  • Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood.; 109:1298-1306 ; Barbash,IM, P Chouraqui, J Baron, M S Feinberg, S Etzion, A Tessone, L Miller, E Guetta, D Zipori, L H Kedes, R A Kloner, J Leor, 2003, Systemic delivery of bone marrow-derived mesenchymal stem cells to the infarcted myocardium: feasibility, cell migration, and body distribution: Circulation, v. 108, p.
  • Intravenous bone marrow stromal cell therapy reduces apoptosis and promotes endogenous cell proliferation after stroke in female rat.
  • J Neurosci Res. Sep 15;73(6):778-86 Colter DC, Sekiya I, Prockop DJ. (2001) Identification of a subpopulation of rapidly self renewing and multipotential adult stem cells in colonies of human marrow stromal cells. Proc Natl Acad Sci USA. 98:7841-7845 ; De Bari C, Dell'Accio F, Vandenabeele F, Vermeesch JR, Raymackers JM, Luyten FP. (2003) Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane. J Cell Biol.
  • Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007 Aug 1;179(3):1855-63 ; Haynesworth SE, Baber MA, Caplan AI. (1992) Cell surface antigens on human marrowderived mesenchymal cells are detected by monoclonal antibodies. Bone.; 13:69-80 ; Hogg JC, Coxson HO, Brumwell ML, Beyers N, Doerschuk CM, MacNee W, Wiggs BR. (1994) Erythrocyte and polymorphonuclear cell transit time and concentration in human pulmonary capillaries.
  • Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury.
  • Proc Natl Acad Sci USA. 104:11002-7 Pereira RF, O'Hara MD, Laptev AV, Halford KW, Pollard MD, Class R, Simon D, Livezey K, Prockop DJ. (1998) Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta.
  • Proc Natl Acad Sci U S A 95:1142-1147 Penolazzi L, Lambertini E, Tavanti E, Torreggiani E, Vesce F, Gambari R, Piva R.
  • Serum deprivation of human marrow stromal cells selects for a sub-population of early progenitor cells with enhanced expression of Oct-4 and other embryonic genes.
  • Prockop DJ. (2007) "Stemness” does not explain the repair of many tissues bymesenchymal stem/multipotent stromal cells (MSCs).
  • TSG-6 tumor necrosis factor-alpha-stimulated gene-6
  • Multipotent stromal cells are activated to reduce apoptosis in part by upregulation and secretion of stanniocalcin-1.
  • TSG-6 potentiates the antitissue kallikrein activity of inter-alpha-inhibitor through bikunin release. Am. J. Respir. Cell Mol. Biol.
  • Hyaluronan binding to link module of TSG-6 and to G1-domain of aggrecan is differently regulated by pH.
  • J. Biol. Chem. Heymans,S., Luttun,A., Nuyens,D., Theilmeier,G., Creemers,E., Moons,L., Dyspersin,G.D., Cleutjens,J.P., Shipley,M., Angellilo,A., Levi,M., Nube,O., Baker,A., Keshet,E., Lupu,F., Herbert,J.M., Smits,J.F., Shapiro,S.D., Baes,M., Borgers,M., Collen,D., Daemen,M.J., and Carmeliet,P.
  • Multipotent human stromal cells improve cardiac function after myocardial infarction in mice without long-term engraftment.
  • Bringing cardiovascular cell-based therapy to clinical application perspectives based on a National Heart, Lung, and Blood Institute Cell Therapy Working Group meeting. Am. Heart J.
  • MMD metachromatic leukodystrophy
  • MRS-IH Hurler syndrome
  • Bone marrow-derived mesenchymal stem cells remain host-derived despite successful hematopoietic engraftment after allogeneic transplantation in patients with lysosomal and peroxisomal storage diseases.
  • Exp Hematol. 27, 1675-1681 Krause,U., Harter,C., Seckinger,A., Wolf,D., Reinhard,A., Bea,F., Dengler,T., Hardt,S., Ho,A., Katus,H.A., Kuecherer,H., and Hansen,A. (2007).
  • Intravenous delivery of autologous mesenchymal stem cells limits infarct size and improves left ventricular function in the infarcted porcine heart.
  • CD34-like protein PODXL and alpha6-integrin identify early progenitor MSCs with increased clonogenicity and migration to infarcted heart in mice. Blood 113, 816-826 ; Lee,T.H., Wisniewski,H.G., and Vilcek,J. (1992).
  • TSG-6 tumor necrosis factor-inducible protein
  • J. Cell Biol. 116, 545-557 Lindsey,M., Wedin,K., Brown,M.D., Keller,C., Evans,A.J., Smolen,J., Burns,A.R., Rossen,R.D., Michael,L., and Entman,M. (2001).
  • Over-expression of CNTF in Mesenchymal Stem Cells reduces demyelination and induces clinical recovery in experimental autoimmune encephalomyelitis mice. J. Neuroimmunol. 206, 58-69 ; McBride,C., Gaupp,D., and Phinney,D.G. (2003). Quantifying levels of transplanted murine and human mesenchymal stem cells in vivo by real-time PCR. Cytotherapy. 5, 7-18 ; McCombe,P.A. and Read,S.J. (2008).
  • TSG-6 a pluripotent inflammatory mediator? Biochem. Soc. Trans. 34, 446-450 ; Mindrescu,C., Dias,A.A.,Olszewski,R.J., Klein,M.J., Reis,L.F., and Wisniewski,H.G. (2002). Reduced susceptibility to collagen-induced arthritis in DBA/1J mice expressing the TSG-6 transgene. Arthritis Rheum.
  • Metalloproteinase inhibitor counters high-energy phosphate depletion and AMP deaminase activity enhancing ventricular diastolic compliance in subacute heart failure. J. Pharmacol. Exp. Ther. 317, 506-513 ; Parr,A.M., Tator,C.H., and Keating,A. (2007).
  • Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant. 40, 609-619 ; Peister,A., Mellad,J.A., Larson.B.L., Hall,B.M., Gibson,L.F., and Prockop,D.J. (2004).
  • MSCs bone marrow
  • Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Rheumatology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Immunology (AREA)
  • General Engineering & Computer Science (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Claims (4)

  1. Utilisation d'une population de cellules souches mésenchymateuses (MSC) purifiées qui a été mise en contact avec un ou plusieurs parmi le TNF-alpha, l'IL-1 et le LPS dans des conditions permettant d'exprimer des taux accrus de protéine TSG-6 dans la fabrication d'une préparation pharmaceutique destinée à réduire un ou plusieurs symptômes d'une nécrose des cellules musculaires cardiaques chez un sujet mammifère.
  2. Utilisation selon la revendication 1, dans laquelle ladite préparation pharmaceutique est destinée à être administrée par une administration intramusculaire dans le muscle cardiaque ou une administration intraveineuse.
  3. Utilisation d'une population de cellules souches mésenchymateuses (MSC) transgéniques qui (a) comprend une séquence de nucléotides hétérologue codant pour une protéine TSG-6, et (b) exprime ladite protéine TSG-6 dans la fabrication d'une préparation pharmaceutique destinée à réduire un ou plusieurs symptômes d'une nécrose des cellules musculaires cardiaques chez un sujet mammifère.
  4. Utilisation selon la revendication 3, dans laquelle ladite préparation pharmaceutique est destinée à être administrée par une administration intramusculaire dans le muscle cardiaque ou une administration intraveineuse.
EP09767083.0A 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque Active EP2294183B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP15200119.4A EP3029061B1 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement de lésions du tissu cardiaque

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US7373908P 2008-06-18 2008-06-18
PCT/US2009/003657 WO2009154770A2 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP15200119.4A Division EP3029061B1 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement de lésions du tissu cardiaque

Publications (3)

Publication Number Publication Date
EP2294183A2 EP2294183A2 (fr) 2011-03-16
EP2294183A4 EP2294183A4 (fr) 2012-08-15
EP2294183B1 true EP2294183B1 (fr) 2015-12-16

Family

ID=41434601

Family Applications (2)

Application Number Title Priority Date Filing Date
EP09767083.0A Active EP2294183B1 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque
EP15200119.4A Active EP3029061B1 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement de lésions du tissu cardiaque

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP15200119.4A Active EP3029061B1 (fr) 2008-06-18 2009-06-18 Cellules souches mésenchymateuses, compositions et procédés pour le traitement de lésions du tissu cardiaque

Country Status (4)

Country Link
US (1) US10035834B2 (fr)
EP (2) EP2294183B1 (fr)
JP (2) JP5866601B2 (fr)
WO (1) WO2009154770A2 (fr)

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9878003B2 (en) 2006-03-06 2018-01-30 The University Of Manchester Method of treating bone disorders using TSG-6
WO2011044251A1 (fr) * 2009-10-06 2011-04-14 The Cohen Mcniece Foundation Préparation et utilisation de cellules stromales pour le traitement d'affections cardiaques
US9139812B2 (en) 2010-02-18 2015-09-22 Kang Stem Holdings Co., Ltd CD49F promoting proliferation, multipotency and reprogramming of adult stem cells through PI3K/AKT/GSK3 pathway
EP2566566B1 (fr) * 2010-05-03 2017-09-06 The Texas A&M University System Cellules souches adultes/cellules progénitrices et protéines de cellules souches pour traiter des maladies et des lésions oculaires
EP2741757B1 (fr) 2011-09-11 2018-05-16 Minovia Therapeutics Ltd. Compositions de mitochondries fonctionnelles et leurs utilisations
EP2626369A1 (fr) * 2011-11-24 2013-08-14 Fundació Institut d'Investigació Biomèdica de Bellvitge (IDIBELL) Cellules souches mésenchymales conçues et leur utilisation thérapeutique
RU2482183C1 (ru) * 2011-12-02 2013-05-20 Государственное научное учреждение Всероссийский научно-исследовательский институт экспериментальной ветеринарии им. Я.Р. Коваленко Российской академии сельскохозяйственных наук (ГНУ ВИЭВ Россельхозакадемии) КУЛЬТУРА МУЛЬТИПОТЕНТНЫХ МЕЗЕНХИМНЫХ СТВОЛОВЫХ КЛЕТОК, ВЫДЕЛЕННЫХ ИЗ КОСТНОГО МОЗГА КРУПНОГО РОГАТОГО СКОТА (Medulla ossium Bos taurus), ДЛЯ ВЕТЕРИНАРИИ, КЛЕТОЧНОЙ И ТКАНЕВОЙ ИНЖЕНЕРИИ
RU2482182C1 (ru) * 2011-12-02 2013-05-20 Государственное научное учреждение Всероссийский научно-исследовательский институт экспериментальной ветеринарии им. Я.Р. Коваленко Российской академии сельскохозяйственных наук (ГНУ ВИЭВ Россельхозакадемии) КУЛЬТУРА МУЛЬТИПОТЕНТНЫХ МЕЗЕНХИМНЫХ СТВОЛОВЫХ КЛЕТОК, ВЫДЕЛЕННЫХ ИЗ ЖИРОВОЙ ТКАНИ КРУПНОГО РОГАТОГО СКОТА (Textus adiposus Bos taurus), ДЛЯ ВЕТЕРИНАРИИ, КЛЕТОЧНОЙ И ТКАНЕВОЙ ИНЖЕНЕРИИ
WO2013171752A1 (fr) 2012-05-16 2013-11-21 Minovia Therapeutics Ltd. Compositions et procédés d'induction de l'angiogenèse
US20150265675A1 (en) * 2012-10-17 2015-09-24 Darwin J. Prockop Treatment of Brain Injury or Trauma with TSG-6 Protein
WO2016081032A2 (fr) * 2014-07-11 2016-05-26 The Texas A&M University System Cellules souches mésenchymateuses issues de cellules souches pluripotentes induites
WO2016018761A1 (fr) * 2014-07-28 2016-02-04 The Texas A&M University System Cellules souches mésenchymateuses exprimant des biomarqueurs permettant de prédire l'efficacité des cellules souches mésenchymateuses pour traiter des maladies et des troubles
JP7015169B2 (ja) 2015-02-26 2022-02-02 ミノヴィア セラピューティクス リミテッド 機能性ミトコンドリアで富化された哺乳動物細胞
KR101816246B1 (ko) 2016-09-07 2018-01-08 에스씨엠생명과학 주식회사 염증 자극된 중간엽 줄기세포를 포함하는 면역질환 또는 염증 질환의 예방 또는 치료용 약학적 조성물
PT3551748T (pt) * 2016-12-12 2022-09-29 Health And Biotech France H&B France Células estaminais do mesênquima derivadas de tecido perinatal: método de preparação e suas utilizações
CA3046743A1 (fr) * 2016-12-16 2018-06-21 Osaka Air Machine Service, Ltd. Agent de cicatrisation des tissus
WO2018123968A1 (fr) * 2016-12-28 2018-07-05 株式会社ツーセル Agent de réparation pour lésion de tissu vivant et procédé de production dudit agent de réparation
CN110423721B (zh) * 2018-05-01 2024-02-27 云南济慈再生医学研究院有限公司 一种年轻化的修复型成纤维细胞的制备方法及其应用
EP3823640A4 (fr) 2018-07-22 2022-05-18 Minovia Therapeutics Ltd. Thérapie d'augmentation mitochondriale de maladies musculaires
WO2021040735A1 (fr) * 2019-08-30 2021-03-04 The Texas A&M University System Compositions de cellules souches mésenchymateuses exemptes de xénogène et méthodes d'utilisation
MX2023000780A (es) * 2020-07-17 2023-02-14 Lg Chemical Ltd Composicion para prevenir o tratar la osteoartritis, que comprende celulas madre mesenquimales que expresan el gen 6 inducible por el factor de necrosis tumoral.

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US5272065A (en) 1983-10-20 1993-12-21 Research Foundation Of State University Of New York Regulation of gene expression by employing translational inhibition of MRNA utilizing interfering complementary MRNA
US4978745A (en) 1987-11-23 1990-12-18 Centocor, Inc. Immunoreactive heterochain antibodies
US4975369A (en) 1988-04-21 1990-12-04 Eli Lilly And Company Recombinant and chimeric KS1/4 antibodies directed against a human adenocarcinoma antigen
US5087617A (en) 1989-02-15 1992-02-11 Board Of Regents, The University Of Texas System Methods and compositions for treatment of cancer using oligonucleotides
EP0567575B1 (fr) * 1991-01-14 1999-10-13 New York University Poteine induite par la cytokine, adn tsg-6 codant pour cette proteine et ses utilisations
CA2112799A1 (fr) 1991-07-03 1993-01-21 Stephen P. Squinto Methode et systeme d'analyse de l'activite des neurotrophines
WO1995021381A1 (fr) 1994-02-01 1995-08-10 The Rockefeller University Procedes et agents de mesure et de regulation de la resistance a plusieurs medicaments anti-cancer
WO1997004075A1 (fr) * 1995-07-20 1997-02-06 New York University Compositions pharmaceutiques contenant la proteine tsg-6 pour traiter des maladies inflammatoires et des pathologies liees au cancer, et procedes de traitement
NZ337445A (en) 1997-02-26 2000-12-22 Gen Hospital Corp Transgenic animals and cell lines for screening drugs effective for the treatment or prevention of alzheimer's disease
CA2296704C (fr) * 1997-07-14 2010-10-19 Osiris Therapeutics, Inc. Regeneration du muscle cardiaque a l'aide de cellules souche mesenchymateuses
ES2314786T3 (es) * 1998-04-02 2009-03-16 Genentech, Inc. Uso de interferon gamma para el tratamiento de la hipertrofia cardiaca.
US20040214836A1 (en) * 1998-05-29 2004-10-28 Cheresh David A. Method of treatment of myocardial infarction
CN1318105A (zh) * 1998-09-15 2001-10-17 M&E生物技术公司 负调节Osteoprotegerin配体活性的方法
US6872546B1 (en) * 1998-12-23 2005-03-29 Human Genome Sciences, Inc. Hyaluronan-binding proteins and encoding genes
US6610503B1 (en) 1999-03-17 2003-08-26 Xenogen Corporation Animal models for predicting sepsis mortality
WO2000077246A2 (fr) 1999-06-14 2000-12-21 Novo Nordisk A/S COMPOSES INHIBANT L'ACTIVITE DU COMPLEXE FVIIa/TF
US6632979B2 (en) 2000-03-16 2003-10-14 Genentech, Inc. Rodent HER2 tumor model
EP1138694A1 (fr) * 2000-03-30 2001-10-04 University Of Manitoba Peptide de type hirulogue
US6905839B2 (en) 2002-07-31 2005-06-14 Takashi Furuta In vivo phenotyping for human cytochrome P450 3A activity
US20070010008A1 (en) * 2005-06-29 2007-01-11 Tissuetech, Inc. Ex vivo expansion of primary animal cells
CA2540135C (fr) * 2003-10-03 2012-12-04 Keiichi Fukuda Methode permettant d'induire la differenciation des cellules souches en cardiomyocytes
US9002545B2 (en) 2011-01-07 2015-04-07 Wabtec Holding Corp. Data improvement system and method

Also Published As

Publication number Publication date
EP2294183A4 (fr) 2012-08-15
WO2009154770A2 (fr) 2009-12-23
EP3029061A1 (fr) 2016-06-08
US10035834B2 (en) 2018-07-31
EP2294183A2 (fr) 2011-03-16
JP5866601B2 (ja) 2016-02-17
EP3029061B1 (fr) 2017-09-13
JP2011525355A (ja) 2011-09-22
WO2009154770A3 (fr) 2010-03-18
JP2015199755A (ja) 2015-11-12
US20100278790A1 (en) 2010-11-04

Similar Documents

Publication Publication Date Title
EP2294183B1 (fr) Cellules souches mésenchymateuses, compositions et procédés pour le traitement des lésions du tissu cardiaque
Lee et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6
Lewis‐McDougall et al. Aged‐senescent cells contribute to impaired heart regeneration
AU2013290146B2 (en) Mesenchymal-like stem cells derived from human embryonic stem cells, methods and uses thereof
CN110088623B (zh) 选择用于治疗免疫病症的高效干细胞的方法
Mehrabani et al. Deferoxamine preconditioning to restore impaired HIF‐1α‐mediated angiogenic mechanisms in adipose‐derived stem cells from STZ‐induced type 1 diabetic rats
US11607428B2 (en) Mesenchymal stem cell-derived extracellular vesicles and uses thereof for treating and diagnosing fibrotic diseases
KR102414662B1 (ko) 향상된 탯줄 유래 부착형 줄기세포, 그의 제조방법 및 용도
Paradisi et al. Human mesenchymal stem cells produce bioactive neurotrophic factors: source, individual variability and differentiation issues
US20230181649A1 (en) Exosomes for disease treatment
EP2970901B1 (fr) Population de cellules souches cardiaques adultes
JP2024515690A (ja) 疾患の治療のための間葉系間質細胞からの細胞外小胞
Cabezas et al. In vitro preconditioning of equine adipose mesenchymal stem cells with prostaglandin E2, substance P and their combination changes the cellular protein secretomics and improves their immunomodulatory competence without compromising stemness
US20230302059A1 (en) Umbilical cord-derived adherent stem cells, preparation method therefor, and use thereof
WO2022033451A1 (fr) Utilisations de msc dans l'amélioration des complications thrombotiques dans une pneumonie de covid-19
US20240150716A1 (en) Method for obtaining tumor-hypoxia educated regenerative macrophages and use thereof in regenerative medicine
Cheung Characterization of DPP4⁺ fibroadipogenic progenitors in skeletal muscle
WO2022212805A2 (fr) Cellules à action paracrine à modulation immunitaire (impacs)
PL242803B1 (pl) Panel ekspresyjny i zastosowanie panelu ekspresyjnego do badań przydatności ludzkich mezenchymalnych komórek macierzystych ADSC do wspomagania regeneracji mięśni szkieletowych oraz sposób oceny przydatności komórek ADSC do wspomagania regeneracji mięśni szkieletowych
Kodály Valsette: pour piano/par Z. Kodály.

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101223

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA RS

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/525 20060101AFI20110427BHEP

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602009035272

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C12N0005000000

Ipc: C07K0014525000

A4 Supplementary search report drawn up and despatched

Effective date: 20120713

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/525 20060101AFI20120709BHEP

Ipc: C12N 5/077 20100101ALI20120709BHEP

17Q First examination report despatched

Effective date: 20130228

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150618

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 765533

Country of ref document: AT

Kind code of ref document: T

Effective date: 20160115

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009035272

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20151216

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160316

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 765533

Country of ref document: AT

Kind code of ref document: T

Effective date: 20151216

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160317

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160416

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20160418

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009035272

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

26N No opposition filed

Effective date: 20160919

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160618

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20090618

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160630

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20160618

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151216

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20220628

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20220627

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DE

Payment date: 20220629

Year of fee payment: 14

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602009035272

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20230618

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20240103

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230618